Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
2.
Pediatr Res ; 89(3): 483-487, 2021 02.
Article in English | MEDLINE | ID: mdl-32299088

ABSTRACT

BACKGROUND: Genetic predispositions in cases suffering sudden unexpected infant death have been a research focus worldwide during the past decade. Despite large efforts, there is still uncertainty concerning the molecular pathogenesis of these deaths. With genetic technology in constant development, the possibility of an alternative approach into this research field has become available, like mRNA expression studies. METHODS: In this study, we investigated mRNA gene expression in 14 cases who died suddenly and unexpectedly from infection without a history of severe illness prior to death. The control group included eight accidents, two cases of natural death, one undetermined, one case of medical malpractice, and two homicides. The study included tissue from liver, heart, and brain using Illumina whole-genome gene expression assay. RESULTS: From the array, 19 genes showed altered expression in the infectious deaths compared to controls. Tissue from the heart showed 15 genes with altered mRNA expression compared to the control group. CONCLUSIONS: Downregulation of KCNE5 in heart tissue from cases of infectious death was of particular interest. Variants of KCNE5 are associated with Brugada syndrome and sudden death and could be responsible for the fatal outcome in the group of infectious death. IMPACT: KCNE5 is downregulated in tissue from the heart in cases of infectious death in infancy. This study provides knowledge about the gene expression profile in cases of infectious death. Variants of a gene known to give increased risk of cardiac arrhythmia is downregulated in cases of infectious death in infancy. The results could give us better knowledge as to why some infants do not survive an infection. This study provides a candidate gene for future studies.


Subject(s)
Bacterial Infections/mortality , Death, Sudden/etiology , RNA, Messenger/biosynthesis , Transcriptome , Virus Diseases/mortality , Bacterial Infections/genetics , Case-Control Studies , Cause of Death , Diagnosis, Differential , Down-Regulation , Female , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Infant , Liver/metabolism , Male , Myocardium/metabolism , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/physiology , Sudden Infant Death/diagnosis , Temporal Lobe/metabolism , Tissue Array Analysis , Virus Diseases/genetics
3.
Forensic Sci Med Pathol ; 15(4): 622-628, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31502215

ABSTRACT

This report details the proceedings and conclusions from the 3rd International Congress on Unexplained Deaths in Infants and Children, held November 26-27, 2018 at the Radcliffe Institute at Harvard University. The Congress was motivated by the increasing rejection of the diagnosis Sudden Infant Death Syndrome (SIDS) in the medical examiner community, leading to falsely depressed reported SIDS rates and undermining the validity and reliability of the diagnosis, which remains a leading cause of infant and child mortality. We describe the diagnostic shift away from SIDS and the practical issues contributing to it. The Congress was attended by major figures and opinion leaders in this area from countries significantly engaged in this problem. Four categories (International Classification of Diseases (ICD)-11 categories of MH11, MH12, MH14, PB00-PB0Z) were recommended for classification, and explicit definitions and guidance were provided for death certifiers. SIDS was reframed as unexplained sudden death in infancy or SIDS/MH11 to emphasize that either term signifies the lack of explanation following a rigorous investigation. A distinct category for children over the age of 1 was recommended (MH12). Definitions and exclusions were provided for the alternative categories of accidental asphyxia and undetermined. As recommended, unexplained sudden death in infancy or SIDS on a death certificate will code a unique, trackable entity, accurately reflecting the inability to determine a definitive explanation, while satisfying surveillance needs and reliable identification for research efforts. The conclusions will be submitted to the World Health Organization for inclusion in the upcoming ICD-11.


Subject(s)
Death, Sudden , Sudden Infant Death/classification , Terminology as Topic , Accidents , Asphyxia , Bedding and Linens , Child , Forensic Medicine , Humans , Infant , International Classification of Diseases
4.
Acta Paediatr ; 108(7): 1262-1266, 2019 07.
Article in English | MEDLINE | ID: mdl-30550627

ABSTRACT

AIM: The aim of this study was to investigate if a range of known rare and common genetic variants in the Toll-like receptor 4 (TLR4)/myeloid differentiation primary response 88 (MyD88) pathway were present or overrepresented in sudden infant death syndrome (SIDS) compared to controls. METHODS: Genetic variations in the genes encoding TLR4, MyD88 and Interleukin-1 receptor-associated kinase 4 were analysed. The subjects investigated included 158 SIDS cases with a median age of 15.25 weeks (2-47 weeks), 80 cases of infectious death with a median age of 24.9 weeks (0-285 weeks) and 199 adult controls with a median age of 50 years (11-86 years). The cases were collected in the years 1988-2017, and the autopsies were performed at the Department of Forensic Sciences at Oslo University Hospital, Oslo, Norway. RESULTS: The results showed that none of the genetic variants selected from the MyD88 pathway were associated with neither SIDS nor infectious death. Most of the rare genetic variants were homozygote for the common allele in all groups, while the rest revealed allelic variation. CONCLUSION: The genetic variations investigated in this study did not appear to be involved in the pathogenesis of SIDS.


Subject(s)
Myeloid Differentiation Factor 88/genetics , Sudden Infant Death/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Myeloid Differentiation Factor 88/metabolism , Polymorphism, Single Nucleotide , Sudden Infant Death/immunology , Young Adult
5.
Acta Paediatr ; 106(9): 1474-1480, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28520217

ABSTRACT

AIM: Disturbances in brain function and development may play a role in sudden infant death syndrome (SIDS). This Norwegian study aimed to test the hypothesis that specific variants of genes involved in water transport and potassium homeostasis would be predisposing factors for SIDS. METHODS: Genetic variation in the genes encoding aquaporin-4 (AQP4), Kir4.1 (KCNJ10) and α-syntrophin was analysed in 171 SIDS cases (62.6% male) with a median age of 15.5 (2-52) weeks and 398 adult controls (70.6% male) with a median age of 44 (11-91) years. All the subjects were Caucasians who were autopsied from 1988 to 2013. RESULTS: The CC genotype of rs72878794 in the AQP4 gene and a combination of the CC genotype in rs17375748, rs1130183, rs12133079 and rs1186688 in KCNJ10 (4xCC) were found to be associated with SIDS. The SIDS cases with the 4xCC SNP combination were younger than the SIDS cases with other genotype combinations (p = 0.006). CONCLUSION: This study indicates that genetic variations in KCNJ10 and AQP4 may be predisposing factors for SIDS. Alterations in the expression of the AQP4/Kir4.1 complex can disrupt water and ion homeostasis, which may influence brain development and facilitate brain oedema formation This may be especially unfavourable during the first weeks of life.


Subject(s)
Aquaporin 4/genetics , Calcium-Binding Proteins/genetics , Membrane Proteins/genetics , Muscle Proteins/genetics , Potassium Channels, Inwardly Rectifying/genetics , Sudden Infant Death/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Child , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Polymorphism, Single Nucleotide , Young Adult
6.
Pediatr Res ; 80(1): 77-84, 2016 07.
Article in English | MEDLINE | ID: mdl-26959483

ABSTRACT

BACKGROUND: A large number of studies have tried to uncover a genetic predisposition for sudden infant death syndrome (SIDS), but there is still uncertainty concerning the pathogenesis of these deaths. The purpose of this study was to investigate mRNA gene expression in SIDS cases and controls, in order to uncover genes that are differentially expressed in the two groups. METHODS: Tissue from brain, heart, and liver from 15 SIDS cases and 15 controls were included in the study, and mRNA expression was determined using the Illumina whole genome gene expression DASL HT assay. RESULTS: Seventeen genes showed significantly altered expression compared to controls, after correction for multiple testing. Three genes involved in the immune system were of particular interest, including the downregulation of MyD88 in tissue from SIDS brains, as well as the downregulation of the genes encoding CCL3 and UNC13 in the liver. CONCLUSION: These findings indicate that there is an altered expression of genes involved in the inflammatory process in a proportion of SIDS cases, which further strengthen the hypothesis that impaired immune response play a role in this syndrome.


Subject(s)
Chemokine CCL3/genetics , Immunologic Deficiency Syndromes/genetics , Myeloid Differentiation Factor 88/genetics , Nerve Tissue Proteins/genetics , Sudden Infant Death/genetics , Case-Control Studies , Child, Preschool , Female , Gene Expression Profiling , Genetic Predisposition to Disease , Humans , Immune System , Infant , Liver/metabolism , Male , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Signal Transduction , Staphylococcus aureus , Streptococcal Infections , Streptococcus pneumoniae , Tissue Distribution
7.
Clin Cancer Res ; 21(16): 3759-70, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25910952

ABSTRACT

PURPOSE: Colorectal cancer has high incidence and mortality worldwide. Patients with microsatellite instable (MSI) tumors have significantly better prognosis than patients with microsatellite stable (MSS) tumors. Considerable variation in disease outcome remains a challenge within each subgroup, and our purpose was to identify biomarkers that improve prediction of colorectal cancer prognosis. EXPERIMENTAL DESIGN: Mutation analyses of 42 MSI target genes were performed in two independent MSI tumor series (n = 209). Markers that were significantly associated with prognosis in the test series were assessed in the validation series, followed by functional and genetic explorations. The clinical potential was further investigated by immunohistochemistry in a population-based colorectal cancer series (n = 903). RESULTS: We identified the cell-cycle gene regulator of chromosome condensation 2 (RCC2) as a cancer biomarker. We found a mutation in the 5' UTR region of RCC2 that in univariate and multivariate analyses was significantly associated with improved outcome in the MSI group. This mutation caused reduction of protein expression in dual luciferase gene reporter assays. siRNA knockdown in MSI colon cancer cells (HCT15) caused reduced cell proliferation, cell-cycle arrest, and increased apoptosis. Massive parallel sequencing revealed few RCC2 mutations in MSS tumors. However, weak RCC2 protein expression was significantly associated with poor prognosis, independent of clinical high-risk parameters, and stratifies clinically important patient subgroups with MSS tumors, including elderly patients (>75 years), stage II patients, and those with rectal cancer. CONCLUSIONS: Impaired RCC2 affects functional and clinical endpoints of colorectal cancer. High-risk patients with either MSI or MSS tumors can be identified with cost-effective routine RCC2 assays.


Subject(s)
Biomarkers, Tumor/genetics , Chromosomal Proteins, Non-Histone/genetics , Colorectal Neoplasms/genetics , Guanine Nucleotide Exchange Factors/genetics , Microsatellite Instability , Age Factors , Aged , Aged, 80 and over , Biomarkers, Tumor/biosynthesis , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/biosynthesis , Chromosomes/genetics , Colorectal Neoplasms/pathology , DNA Mutational Analysis , Disease-Free Survival , Female , Guanine Nucleotide Exchange Factors/biosynthesis , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis
8.
Int J Cancer ; 136(4): 844-53, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-24948044

ABSTRACT

We have previously shown that gastrointestinal cancers display similar epigenetic aberrations. In a recent study, we identified frequently methylated genes for cholangiocarcinoma (CDO1, DCLK1, SFRP1 and ZSCAN18), where one of these genes, DCLK1, was also confirmed to be highly methylated in colorectal cancer. The aim of the present study was to determine whether these four genes, in addition to one gene found to be methylated in colon cancer cell lines (ZNF331), are commonly methylated across gastrointestinal malignancies, as well as explore their role as potential biomarkers. Quantitative methylation specific PCR (qMSP) of colorectal cancer (n=164) and normal colorectal mucosa (n=106) samples showed that all genes were frequently methylated in colorectal cancer (71-92%) with little or no methylation in normal mucosa (0-3%). Methylation of minimum two of these five genes identified 95% of the tumors with a specificity of 98%, and an area under the receiver operating characteristics curve (AUC) of 0.98. For gastric (n=25) and pancreatic (n=20) cancer, the same panel detected 92% and 90% of the tumors, respectively. Seventy-four cancer cell lines were further analyzed by qMSP and real time RT-PCR. In addition to the previously reported DCLK1, a high negative correlation between promoter DNA methylation and gene expression was observed for CDO1, ZNF331 and ZSCAN18. In conclusion, the high methylation frequency of these genes in colorectal- as well as in gastric-, pancreatic- and bile duct cancer confirmed an epigenetic similarity between gastrointestinal cancer types, and simultaneously demonstrated their potential as biomarkers, particularly for colorectal cancer detection.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Cysteine Dioxygenase/genetics , DNA Methylation , DNA-Binding Proteins/genetics , Neoplasm Proteins/genetics , Adult , Aged , Aged, 80 and over , Area Under Curve , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Bile Ducts, Intrahepatic/metabolism , Biomarkers, Tumor/metabolism , Case-Control Studies , Cell Line, Tumor , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Colorectal Neoplasms/metabolism , Cysteine Dioxygenase/metabolism , DNA-Binding Proteins/metabolism , Gene Expression , Humans , Middle Aged , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Promoter Regions, Genetic , ROC Curve , Sequence Analysis, DNA , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Young Adult
9.
Int J Cancer ; 135(9): 2077-84, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-24687856

ABSTRACT

Colorectal cancer is a global health challenge with high incidence rate and mortality. The patients' prognosis is strongly associated with disease stage and currently there is a need for improved prognostic and predictive biomarkers. In this study, novel colorectal cancer-specific transcript structures were nominated from whole transcriptome sequencing of seven colorectal cancer cell lines, two primary colorectal carcinomas with corresponding normal colonic mucosa and 16 normal tissues. The nominated transcripts were combined with gene level outlier expression analyses in a cohort of 505 colorectal cancers to identify biomarkers with capacity to stratify colorectal cancer subgroups. The transcriptome sequencing data and outlier expression analysis revealed 11 novel colorectal cancer-specific exon-exon junctions, of which 3 were located in the gene VNN1. The junctions within VNN1 were further characterized using rapid amplification of cDNA ends (RACE) and the prevalence of the subsequently characterized novel transcript, VNN1-AB, was investigated by real-time RT-PCR in 291 samples of miscellaneous origins. VNN1-AB was not present in any of the 43 normal colorectal tissue samples investigated, but in 5 of the 6 polyps, and 102 of the 136 (75%) colorectal cancers. We have identified a novel transcript of the VNN1 gene, with an organ-confined complete specificity for colorectal neoplasia.


Subject(s)
Adenoma/genetics , Alternative Splicing/genetics , Amidohydrolases/genetics , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Exons/genetics , Adenoma/pathology , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/pathology , GPI-Linked Proteins/genetics , Gene Expression Profiling , Humans , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
10.
PLoS One ; 9(3): e91264, 2014.
Article in English | MEDLINE | ID: mdl-24608966

ABSTRACT

VTI1A-TCF7L2 was reported as a recurrent fusion gene in colorectal cancer (CRC), found to be expressed in three out of 97 primary cancers, and one cell line, NCI-H508, where a genomic deletion joins the two genes [1]. To investigate this fusion further, we analyzed high-throughput DNA and RNA sequencing data from seven CRC cell lines, and identified the gene RP11-57H14.3 (ENSG00000225292) as a novel fusion partner for TCF7L2. The fusion was discovered from both genome and transcriptome data in the HCT116 cell line. By triplicate nested RT-PCR, we tested both the novel fusion transcript and VTI1A-TCF7L2 for expression in a series of 106 CRC tissues, 21 CRC cell lines, 14 normal colonic mucosa, and 20 normal tissues from miscellaneous anatomical sites. Altogether, 42% and 45% of the CRC samples expressed VTI1A-TCF7L2 and TCF7L2-RP11-57H14.3 fusion transcripts, respectively. The fusion transcripts were both seen in 29% of the normal colonic mucosa samples, and in 25% and 75% of the tested normal tissues from other organs, revealing that the TCF7L2 fusion transcripts are neither specific to cancer nor to the colon and rectum. Seven different splice variants were detected for the VTI1A-TCF7L2 fusion, of which three are novel. Four different splice variants were detected for the TCF7L2-RP11-57H14.3 fusion. In conclusion, we have identified novel variants of VTI1A-TCF7L2 fusion transcripts, including a novel fusion partner gene, RP11-57H14.3, and demonstrated detectable levels in a large fraction of CRC samples, as well as in normal colonic mucosa and other tissue types. We suggest that the fusion transcripts observed in a high frequency of samples are transcription induced chimeras that are expressed at low levels in most samples. The similar fusion transcripts induced by genomic rearrangements observed in individual cancer cell lines may yet have oncogenic potential as suggested in the original study by Bass et al.


Subject(s)
Colorectal Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , RNA Splicing/genetics , Transcription Factor 7-Like 2 Protein/genetics , Base Sequence , Exons/genetics , Gene Rearrangement/genetics , Genetic Loci , Genome, Human/genetics , HCT116 Cells , Humans , Molecular Sequence Data , RNA Splice Sites/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Transcription Factor 7-Like 2 Protein/metabolism
11.
Acta Paediatr ; 103(4): 393-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24286237

ABSTRACT

AIM: The purpose of this study was to investigate common polymorphisms in the genes encoding monoamine oxidase A (MAOA) and serotonin transporter (5-HTT) in Norwegian cases of sudden infant death syndrome (SIDS). This was done to further elucidate the role of genetic variation in these genes and SIDS. METHODS: A variable number of tandem repeat area in the promoter of the MAOA gene and rs25531 in the promoter region of the gene encoding 5-HTT were investigated in 193 SIDS cases and 335 controls. The methods used were polymerase chain reaction, restriction fragment analysis and gel electrophoresis. RESULTS: There were no differences between SIDS cases and controls for any of the investigated polymorphisms. This was also true when male and female SIDS cases were analysed separately. CONCLUSION: This article indicates that neither the VNTR in the promoter of the MAOA gene, nor rs25531 in the gene encoding 5-HTT, is involved in SIDS. However, as medullary serotonergic abnormalities most likely contribute to the death in at least some SIDS cases, it is important to investigate these genes, as well as other genes involved in the serotonergic network, in more detail.


Subject(s)
Genetic Variation , Monoamine Oxidase/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Sudden Infant Death/genetics , Child, Preschool , Female , Genotype , Humans , Infant , Infant, Newborn , Male , Minisatellite Repeats , Polymorphism, Genetic , Promoter Regions, Genetic
13.
Acta Paediatr ; 102(3): 308-13, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23186119

ABSTRACT

AIM: The mucosal immune system and cytokines are activated in a large proportion of cases of sudden infant death syndrome (SIDS). Our aim was to search for a possible association between cytokine polymorphisms and immune stimulation of the laryngeal mucosal in SIDS. METHODS: HLA-DR expression in laryngeal mucosal glands and surface epithelium in 97 SIDS victims was evaluated applying a semi-quantitative scoring system. The findings were related to cytokine gene polymorphisms as well as to the level of various cytokines in the cerebrospinal fluid (CSF). A risk score was established: a score of 0 prepresenting negative HLA-DR, supine position and no fever prior to death. RESULTS: The IL-6 -176CG/CC genotype was found in 92.3% of the SIDS cases with positive score for all risk factors (p = 0.01). Infants with high HLA-DR score had high levels of IL-6 in the cerebrospinal fluid (>30 µg/L) (p = 0.005). Furthermore, the IL-8 SNPs -781 CT/TT genotypes and -251 AA/AT genotypes were observed in 93% of the SIDS cases with one or more of the risk factors present compared with SIDS cases no risk factors reported (p = 0.003 and p = 0.016, respectively). CONCLUSION: This study adds further evidence to the hypothesis that there are genetically associated disturbances of immunological homoeostasis in SIDS.


Subject(s)
HLA-DR Antigens/metabolism , Interleukins/genetics , Laryngeal Mucosa/metabolism , Sudden Infant Death/etiology , Cohort Studies , Female , Humans , Infant , Infant, Newborn , Interleukins/cerebrospinal fluid , Laryngeal Mucosa/pathology , Male , Norway , Polymorphism, Single Nucleotide , Prone Position , Risk Factors , Sudden Infant Death/cerebrospinal fluid , Sudden Infant Death/pathology
14.
Forensic Sci Med Pathol ; 8(4): 414-25, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22941540

ABSTRACT

Reported here are the proceedings of a symposium given in honor of Dr. Henry F. Krous upon his retirement as Clinical Professor of Pathology and Pediatrics at the University of California Schools of Medicine, and as Director of the San Diego SIDS/SUDC Research Project. Dr. Krous' distinguished 37-year-career was dedicated to research into sudden unexpected death in infancy and childhood, notably the sudden infant death syndrome (SIDS) and sudden unexplained death in childhood (SUDC). The presentations were given at the International Conference on Stillbirth, SIDS, and infant survival on October 5, 2012, in Baltimore, MD, USA. Eight colleagues of Dr. Krous whose own professional careers were touched by his efforts discussed forensic issues related to SIDS, tissue banking, animal models in SIDS, brainstem studies in SIDS, genetic studies in SIDS, establishment of a SUDC registry, neuropathologic research in SUDC, and potential shared mechanisms underlying sudden and unexpected death in early life. The wide scope of the presentations crossed the disciplines of forensic pathology, pediatric pathology, neuropathology, neuroscience, physiology, genetics, and bereavement, and attest to Dr. Krous' far-reaching influence upon SIDS and SUDC research.


Subject(s)
Sudden Infant Death/etiology , Sudden Infant Death/pathology , Animals , Autopsy/standards , Biomedical Research , Brain Stem/abnormalities , Brain Stem/pathology , Cell Death , Congresses as Topic , Epilepsy/complications , Forensic Medicine/standards , Humans , Hypoxia-Ischemia, Brain/pathology , Infant , Models, Animal , Neuroglia/pathology , Neurons/pathology , Registries , Tissue Banks
15.
Clin Cancer Res ; 18(21): 6001-10, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22991413

ABSTRACT

PURPOSE: Improved prognostic stratification of patients with stage II and III colorectal cancer is warranted for postoperative clinical decision making. This study was conducted to develop a clinically feasible and robust prognostic classifier for these patients independent of adjuvant treatment. EXPERIMENTAL DESIGN: Global gene expression profiles from altogether 387 stage II and III colorectal cancer tissue samples from three independent patient series were included in the study. ColoGuidePro, a seven-gene prognostic classifier, was developed from a selected Norwegian learning series (n = 95; no adjuvant treatment) using lasso-penalized multivariate survival modeling with cross-validation. RESULTS: The expression signature significantly stratified patients in a consecutive Norwegian test series, in which patients were treated according to current standards [HR, 2.9 (1.1-7.5); P = 0.03; n = 77] and an external validation series [HR, 3.7 (2.0-6.8); P < 0.001; n = 215] according to survival. ColoGuidePro was also an independent predictor of prognosis in multivariate models including tumor stage in both series (HR, ≥ 3.1; P ≤ 0.03). In the validation series, which consisted of patients from other populations (United States and Australia), 5-year relapse-free survival was significantly predicted for stage III patients only (P < 0.001; n = 107). Here, prognostic stratification was independent of adjuvant treatment (P = 0.001). CONCLUSIONS: We present ColoGuidePro, a prognostic classifier developed for patients with stage II and III colorectal cancer. The test is suitable for transfer to clinical use and has best prognostic prediction potential for stage III patients.


Subject(s)
Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Gene Expression Profiling/methods , Transcriptome , Aged , Aged, 80 and over , Colorectal Neoplasms/mortality , Computational Biology , Female , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , Workflow
16.
Clin Transl Gastroenterol ; 3: e27, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-23324654

ABSTRACT

OBJECTIVES: We recently identified a six-gene methylation-based biomarker panel suitable for early detection of colorectal cancer (CRC). In this study, we compared the performance of this novel epi-panel with that of previously identified DNA methylation markers in the same clinical tissue sample sets. METHODS: Quantitative methylation-specific PCR was used to analyze the promoter region of SEPT9 and VIM in a total of 485 tissue samples, divided into test and validation sets. ITGA4, NTRK2, OSMR, and TUBG2 were also included in the analyses. Receiver operating characteristic (ROC) curves were used to compare the performances of the individual biomarkers with that of the novel epi-panel. RESULTS: SEPT9 and VIM were methylated in 82 and 67% of CRCs (n=169) and in 88 and 54% of the adenomas (n=104). Only 3% of the normal mucosa samples (n=107) were methylated for these genes, confirming that the methylation was highly cancer-specific. Areas under the ROC curve (AUC), distinguishing CRCs from normal mucosa, were 0.94 for SEPT9 and 0.81 for VIM. AUC values for separating adenomas from normal mucosa samples were 0.96 and 0.81 for the same genes. In comparison, the novel epi-panel achieved an AUC of 0.98 (CRC) and 0.97 (adenomas).ITGA4, OSMR, NTRK2, and TUBG2 were methylated in 90, 78, 7, and 1% of the CRCs, and in 76, 77, 3, and 0% of the adenomas. Between 0 and 2% of the normal mucosa samples were methylated for the same genes. ITGA4 and OSMR achieved an AUC of 0.96 and 0.92 (CRC vs. normal mucosa), and 0.93 and 0.92 (adenomas vs. normal mucosa). CONCLUSIONS: We have confirmed the high performance of some of the previously identified DNA methylation markers. Furthermore, we showed that a recently reported epi-panel performed better than the individual DNA methylation biomarkers when analyzed in the same tissue samples. This observation was also true for VIM and SEPT9, which are included in commercially available noninvasive tests for CRC. These results further underscore the value of combining a manageable number of individual markers into a panel, which in addition to having a higher sensitivity and specificity might provide a more profound robustness to a noninvasive test compared with single markers.

17.
Int J Cancer ; 131(6): 1479-85, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22173985

ABSTRACT

An alternative transcript variant of SLC39A14, caused by pre-mRNA splicing, was recently suggested as a biomarker for colorectal cancer (CRC). In our study, we have validated the cancer-specific splicing pattern of the mutually exclusive exons 4A and 4B in altogether 244 colorectal tissue samples. Exon-specific quantitative RT-PCR analyses across 136 Stage I-IV CRC samples and 44 normal colonic mucosa samples showed complete cancer-specificity, as well as 94% sensitivity of SLC39A14-exon4B relative to SLC39A14-exon4A expression. However, across 20 samples from a range of healthy tissues, 18 expressed the CRC variant. This was true also for ten benign lymph nodes, demonstrating that the cancer-specificity is mainly confined to the colon and rectum. Hence, clinical use of SLC39A14-exon4B as a detection marker for CRC other than in samples taken from the bowel wall is diminished. Prognostic value by detection of metastasis to lymph nodes is also abated, elucidating an important pitfall to biomarker discovery. However, analyses of ten nondysplastic biopsies from patients with active inflammatory bowel disease showed negative results in seven samples and only weakly positive results in three samples, suggesting value of SLC39A14-exon4B as a marker to distinguish CRC from other pathologic conditions of the colon. In conclusion, the SLC39A14-exon4B transcript variant is a CRC biomarker with high sensitivity and organ-confined specificity. Further use of the transcript and its encoded protein isoform should be explored in an organ-confined context.


Subject(s)
Cation Transport Proteins/genetics , Colorectal Neoplasms/genetics , Exons , Biomarkers, Tumor , Humans , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
18.
Mol Cancer ; 10: 85, 2011 Jul 21.
Article in English | MEDLINE | ID: mdl-21777459

ABSTRACT

BACKGROUND: The presence of cancer-specific DNA methylation patterns in epithelial colorectal cells in human feces provides the prospect of a simple, non-invasive screening test for colorectal cancer and its precursor, the adenoma. This study investigates a panel of epigenetic markers for the detection of colorectal cancer and adenomas. METHODS: Candidate biomarkers were subjected to quantitative methylation analysis in test sets of tissue samples from colorectal cancers, adenomas, and normal colonic mucosa. All findings were verified in independent clinical validation series. A total of 523 human samples were included in the study. Receiver operating characteristic (ROC) curve analysis was used to evaluate the performance of the biomarker panel. RESULTS: Promoter hypermethylation of the genes CNRIP1, FBN1, INA, MAL, SNCA, and SPG20 was frequent in both colorectal cancers (65-94%) and adenomas (35-91%), whereas normal mucosa samples were rarely (0-5%) methylated. The combined sensitivity of at least two positives among the six markers was 94% for colorectal cancers and 93% for adenoma samples, with a specificity of 98%. The resulting areas under the ROC curve were 0.984 for cancers and 0.968 for adenomas versus normal mucosa. CONCLUSIONS: The novel epigenetic marker panel shows very high sensitivity and specificity for both colorectal cancers and adenomas. Our findings suggest this biomarker panel to be highly suitable for early tumor detection.


Subject(s)
Adenoma/diagnosis , Adenoma/genetics , Biomarkers, Tumor/isolation & purification , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Epigenesis, Genetic/genetics , Adenoma/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Carcinoma/diagnosis , Carcinoma/genetics , Carcinoma/pathology , Colorectal Neoplasms/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Intestinal Mucosa/chemistry , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Middle Aged , Sensitivity and Specificity
19.
Genome Med ; 3(5): 32, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21619627

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is a heterogeneous disease that, on the molecular level, can be characterized by inherent genomic instabilities; chromosome instability and microsatellite instability. In the present study we analyze genome-wide disruption of pre-mRNA splicing, and propose transcriptome instability as a characteristic that is analogous to genomic instability on the transcriptome level. METHODS: Exon microarray profiles from two independent series including a total of 160 CRCs were investigated for their relative amounts of exon usage differences. Each exon in each sample was assigned an alternative splicing score calculated by the FIRMA algorithm. Amounts of deviating exon usage per sample were derived from exons with extreme splicing scores. RESULTS: There was great heterogeneity within both series in terms of sample-wise amounts of deviating exon usage. This was strongly associated with the expression levels of approximately half of 280 splicing factors (54% and 48% of splicing factors were significantly correlated to deviating exon usage amounts in the two series). Samples with high or low amounts of deviating exon usage, associated with overall transcriptome instability, were almost completely separated into their respective groups by hierarchical clustering analysis of splicing factor expression levels in both sample series. Samples showing a preferential tendency towards deviating exon skipping or inclusion were associated with skewed transcriptome instability. There were significant associations between transcriptome instability and reduced patient survival in both sample series. In the test series, patients with skewed transcriptome instability showed the strongest prognostic association (P = 0.001), while a combination of the two characteristics showed the strongest association with poor survival in the validation series (P = 0.03). CONCLUSIONS: We have described transcriptome instability as a characteristic of CRC. This transcriptome instability has associations with splicing factor expression levels and poor patient survival.

20.
Epigenetics ; 6(5): 602-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21406965

ABSTRACT

Gap junctions are specialized plasma membrane domains consisting of channels formed by members of the connexin protein family. Gap junctional intercellular communication is often lost in cancers due to aberrant localization or downregulation of connexins, and connexins are therefore suggested to act as tumor suppressor genes in various tissues. The aim of this study was to investigate the expression pattern and DNA promoter methylation status of connexins in colorectal cancer. Expression of six (GJA1, GJA9, GJB1, GJB2, GJC1 and GJD3) connexin genes was detected in normal colonic tissue samples. GJC1 expression was reduced in colorectal carcinomas compared to normal tissue samples. All analyzed connexins were hypermethylated in colon cancer cell lines, although at various frequencies. GJA4, GJB6 and GJD2 were hypermethylated in 60% (29/48), 25% (12/48) and 96% (46/48) of primary colorectal carcinomas, respectively. However, the methylation status was not associated with gene expression. GJC1 has two alternative promoters, which were methylated in 42% (32/76) and 38% (25/65) of colorectal tumors, and in none of the normal mucosa samples. Expression of GJC1 was significantly lower in methylated compared with unmethylated samples (p < 0.01) and was restored in cell lines treated with the demethylating drug 5-aza-2'deoxycytidine. Taken together, DNA hypermethylation of the promoter region of GJC1, encoding connexin45, is an important mechanism in silencing gene expression in colorectal cancer.


Subject(s)
Colorectal Neoplasms/genetics , Connexins/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , Promoter Regions, Genetic/genetics , Adult , Aged , Aged, 80 and over , Cell Communication/genetics , Cell Line, Tumor , Connexin 26 , CpG Islands/genetics , Epigenomics , Female , Gap Junctions/genetics , Gene Silencing , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...